Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Complex networks orchestrate epithelial–mesenchymal transitions

Key Points

  • Epithelial and mesenchymal cells have distinct characteristics. These cell types can be partially or fully interconverted through the processes of epithelial–mesenchymal transition (EMT) and mesenchymal–epithelial transition (MET).

  • EMT is controlled by various extracellular triggers. The intracellular pathways that are activated by these triggers exhibit extensive crosstalk and have many common endpoints.

  • Biophysics as well as cell- and molecular-biology approaches have been combined to provide many novel insights into the process of EMT.

  • EMT has an important role in many embryological processes. Examples include gastrulation, neural-crest development and heart-valve formation.

  • There is mounting evidence that EMT processes are involved in several pathological processes, including would healing, fibrosis and cancer.

  • Better model systems and the identification of genes that mark specific EMT events are required for further progress in this field.

Abstract

Epithelial–mesenchymal transition is an indispensable mechanism during morphogenesis, as without mesenchymal cells, tissues and organs will never be formed. However, epithelial-cell plasticity, coupled to the transient or permanent formation of mesenchyme, goes far beyond the problem of cell-lineage segregation. Understanding how mesenchymal cells arise from an epithelial default status will also have a strong impact in unravelling the mechanisms that control fibrosis and cancer progression.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Images of EMT.
Figure 2: The cycle of epithelial-cell plasticity.
Figure 3: Overview of the molecular networks that regulate EMT.
Figure 4: Molecular events in junctional complexes during EMT.
Figure 5: Tissue remodelling and EMT.
Figure 6: Different signalling pathways cooperate to regulate EMT during neural-crest determination and segregation.

Similar content being viewed by others

References

  1. Schock, F. & Perrimon, N. Molecular mechanisms of epithelial morphogenesis. Annu. Rev. Cell Dev. Biol. 18, 463–493 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Thompson, E. W., Newgreen, D. F. & Tarin, D. Carcinoma invasion and metastasis: a role for epithelial–mesenchymal transition? Cancer Res. 65, 5991–5995 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Friedl, P. Prespecification and plasticity: shifting mechanisms of cell migration. Curr. Opin. Cell Biol. 16, 14–23 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Christ, B. & Ordahl, C. P. Early stages of chick somite development. Anat. Embryol. (Berl) 191, 381–396 (1995).

    Article  CAS  Google Scholar 

  5. Funayama, N., Sato, Y., Matsumoto, K., Ogura, T. & Takahashi, Y. Coelom formation: binary decision of the lateral plate mesoderm is controlled by the ectoderm. Development 126, 4129–4138 (1999).

    CAS  PubMed  Google Scholar 

  6. Locascio, A. & Nieto, M. A. Cell movements during vertebrate development: integrated tissue behaviour versus individual cell migration. Curr. Opin. Genet. Dev. 11, 464–469 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Nieto, M. A. The snail superfamily of zinc-finger transcription factors. Nature Rev. Mol. Cell Biol. 3, 155–166 (2002).

    Article  CAS  Google Scholar 

  8. Gilbert S. F. Developmental Biology, 7th edn (Sinauer Associates Inc., 2003).

    Google Scholar 

  9. Gershengorn, M. C., et al. Epithelial-to-mesenchymal transition generates proliferative human islet precursor cells. Science 306, 2261–2264 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. Radisky, D. C. Epithelial–mesenchymal transition. J. Cell Sci. 118, 4325–4326 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Peinado, H., Portillo, F. & Cano, A. Transcriptional regulation of cadherins during development and carcinogenesis. Int. J. Dev. Biol. 48, 365–375 (2004).

    Article  CAS  PubMed  Google Scholar 

  12. Wang, J. et al. Atrioventricular cushion transformation is mediated by ALK2 in the developing mouse heart. Dev. Biol. 286, 299–310 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zavadil, J. & Bottinger, E. P. TGF-β and epithelial-to-mesenchymal transitions. Oncogene 24, 5764–5774 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Savagner, P. Leaving the neighborhood: molecular mechanisms involved during epithelial–mesenchymal transition. Bioessays 23, 912–923 (2001).

    Article  CAS  PubMed  Google Scholar 

  15. Thiery, J. P. Epithelial–mesenchymal transitions in tumour progression. Nature Rev. Cancer 2, 442–454 (2002). A comprehensive review of the basics of EMT.

    Article  CAS  Google Scholar 

  16. Thiery, J. P. Epithelial–mesenchymal transitions in development and pathologies. Curr. Opin. Cell Biol. 15, 740–746 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Zoltan-Jones, A., Huang, L., Ghatak, S. & Toole, B. P. Elevated hyaluronan production induces mesenchymal and transformed properties in epithelial cells. J. Biol. Chem. 278, 45801–45810 (2003).

    Article  CAS  PubMed  Google Scholar 

  18. Barrallo-Gimeno, A. & Nieto, M. A. The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development 132, 3151–3161 (2005). An excellent review of the different facets of the snail genes.

    Article  CAS  PubMed  Google Scholar 

  19. Polakis, P. Wnt signaling and cancer. Genes Dev. 14, 1837–1851 (2000).

    CAS  PubMed  Google Scholar 

  20. Grille, S. J. et al. The protein kinase Akt induces epithelial mesenchymal transition and promotes enhanced motility and invasiveness of squamous cell carcinoma lines. Cancer Res. 63, 2172–2178 (2003).

    CAS  PubMed  Google Scholar 

  21. Beals, C. R., Sheridan, C. M., Turck, C. W., Gardner, P. & Crabtree, G. R. Nuclear export of NF-ATc enhanced by glycogen synthase kinase-3. Science 275, 1930–1934 (1997).

    Article  CAS  PubMed  Google Scholar 

  22. Zhou, B. P. et al. Dual regulation of Snail by GSK-3β-mediated phosphorylation in control of epithelial–mesenchymal transition. Nature Cell Biol. 6, 931–940 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. Yang, Z. et al. Pak1 phosphorylation of snail, a master regulator of epithelial-to-mesenchyme transition, modulates snail's subcellular localization and functions. Cancer Res. 65, 3179–3184 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Bhowmick, N. A., Zent, R., Ghiassi, M., McDonnell, M. & Moses, H. L. Integrin β1 signaling is necessary for transforming growth factor-β activation of p38MAPK and epithelial plasticity. J. Biol. Chem. 276, 46707–46713 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Prunier, C. & Howe, P. H. Disabled-2 (Dab2) is required for transforming growth factor β-induced epithelial to mesenchymal transition (EMT). J. Biol. Chem. 280, 17540–17548 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Valles, A. M., Boyer, B., Tarone, G. & Thiery, J. P. α2 β1 integrin is required for the collagen and FGF-1 induced cell dispersion in a rat bladder carcinoma cell line. Cell Adhes. Commun. 4, 187–199 (1996).

    Article  CAS  PubMed  Google Scholar 

  27. Balzac, F. et al. E-cadherin endocytosis regulates the activity of Rap1: a traffic light GTPase at the crossroads between cadherin and integrin function. J. Cell Sci. 118, 4765–4783 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Oloumi, A., McPhee, T. & Dedhar, S. Regulation of E-cadherin expression and β-catenin/Tcf transcriptional activity by the integrin-linked kinase. Biochem. Biophys. Acta 1691, 1–15 (2004).

    Article  CAS  PubMed  Google Scholar 

  29. Li, Y., Yang, J., Dai, C., Wu, C. & Liu, Y. Role for integrin-linked kinase in mediating tubular epithelial to mesenchymal transition and renal interstitial fibrogenesis. J. Clin. Invest. 112, 503–516 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gimond, C. et al. Induction of cell scattering by expression of β1 integrins in β1-deficient epithelial cells requires activation of members of the rho family of GTPases and downregulation of cadherin and catenin function. J. Cell Biol. 147, 1325–1340 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Avizienyte, E. & Frame, M. C. Src and FAK signalling controls adhesion fate and the epithelial-to-mesenchymal transition. Curr. Opin. Cell Biol. 17, 542–547 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Yano, H. et al. Roles played by a subset of integrin signaling molecules in cadherin-based cell–cell adhesion. J. Cell Biol. 166, 283–295 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Chu, Y. S. et al. Force measurements in E-cadherin-mediated cell doublets reveal rapid adhesion strengthened by actin cytoskeleton remodeling through Rac and Cdc42. J. Cell Biol. 167, 1183–1194 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Perez-Moreno, M., Jamora, C. & Fuchs, E. Sticky business: orchestrating cellular signals at adherens junctions. Cell 112, 535–548 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Ebnet, K., Suzuki, A., Ohno, S. & Vestweber, D. Junctional adhesion molecules (JAMs): more molecules with dual functions? J. Cell Sci. 117, 19–29 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Takai, Y. & Nakanishi, H. Nectin and afadin: novel organizers of intercellular junctions. J. Cell Sci. 116, 17–27 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Martinez-Rico, C. et al. Separation force measurements reveal different types of modulation of E-cadherin-based adhesion by nectin-1 and-3. J. Biol. Chem. 280, 4753–4760 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Chu, Y. S. et al. Prototypical type-I E-cadherin and type-II cadherin-7 mediate very distinct adhesiveness through their extracellular domain. J. Biol. Chem. Oct 16 2005 (10.1074/jbc.M506185200).

  39. Barrios-Rodiles, M. et al. High-throughput mapping of a dynamic signaling network in mammalian cells. Science 307, 1621–1625 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Ozdamar, B. et al. Regulation of the polarity protein Par6 by TGFβ receptors controls epithelial cell plasticity. Science 307, 1603–1609 (2005).

    Article  CAS  PubMed  Google Scholar 

  41. Wang, H. R. et al. Regulation of cell polarity and protrusion formation by targeting RhoA for degradation. Science 302, 1775–1779 (2003). Exploited an advanced technique to discover new critical partners in tight junctions that couple signalling and morphogenesis.

    Article  CAS  PubMed  Google Scholar 

  42. Fernandez-Serra, M., Consales, C., Livigni, A. & Arnone, M. I. Role of the ERK-mediated signaling pathway in mesenchyme formation and differentiation in the sea urchin embryo. Dev. Biol. 268, 384–402 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Rottinger, E., Besnardeau, L. & Lepage, T. A Raf/MEK/ERK signaling pathway is required for development of the sea urchin embryo micromere lineage through phosphorylation of the transcription factor Ets. Development 131, 1075–1087 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. Oda, H., Tsukita, S. & Takeichi, M. Dynamic behavior of the cadherin-based cell–cell adhesion system during Drosophila gastrulation. Dev. Biol. 203, 435–450 (1998).

    Article  CAS  PubMed  Google Scholar 

  45. Smallhorn, M., Murray, M. J. & Saint, R. The epithelial–mesenchymal transition of the Drosophila mesoderm requires the Rho GTP exchange factor Pebble. Development 131, 2641–2651 (2004).

    Article  CAS  PubMed  Google Scholar 

  46. Yamashita, S. et al. Zinc transporter LIVI controls epithelial–mesenchymal transition in zebrafish gastrula organizer. Nature 429, 298–302 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Ciruna, B. & Rossant, J. FGF signaling regulates mesoderm cell fate specification and morphogenetic movement at the primitive streak. Dev. Cell 1, 37–49 (2001).

    Article  CAS  PubMed  Google Scholar 

  48. Carver, E. A., Jiang, R., Lan, Y., Oram, K. F. & Gridley, T. The mouse snail gene encodes a key regulator of the epithelial–mesenchymal transition. Mol. Cell Biol. 21, 8184–8188 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kemler, R. et al. Stabilization of β-catenin in the mouse zygote leads to premature epithelial–mesenchymal transition in the epiblast. Development 131, 5817–5824 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Huelsken, J. et al. Requirement for β-catenin in anterior–posterior axis formation in mice. J. Cell Biol. 148, 567–578 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lickert, H. et al. Formation of multiple hearts in mice following deletion of β-catenin in the embryonic endoderm. Dev. Cell 3, 171–181 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Meulemans, D. & Bronner-Fraser, M. Gene-regulatory interactions in neural crest evolution and development. Dev. Cell 7, 291–299 (2004). A comprehensive review that provides a basis for stimulating new evolution and development studies.

    Article  CAS  PubMed  Google Scholar 

  53. del Barrio, M. G. & Nieto, M. A. Overexpression of Snail family members highlights their ability to promote chick neural crest formation. Development 129, 1583–1593 (2002).

    CAS  PubMed  Google Scholar 

  54. Vallin, J. et al. Cloning and characterization of three Xenopus slug promoters reveal direct regulation by Lef/β-catenin signaling. J. Biol. Chem. 276, 30350–30358 (2001).

    Article  CAS  PubMed  Google Scholar 

  55. Cheung, M. et al. The transcriptional control of trunk neural crest induction, survival, and delamination. Dev. Cell 8, 179–192 (2005). An advanced study that addresses the complexity of signalling pathways in neural-crest ontogeny in vivo.

    Article  CAS  PubMed  Google Scholar 

  56. Morales, A. V., Barbas, J. A. & Nieto, M. A. How to become neural crest: from segregation to delamination. Semin. Cell Dev. Biol. 16, 655–662 (2005).

    Article  CAS  PubMed  Google Scholar 

  57. Peinado, H., Quintanilla, M. & Cano, A. Transforming growth factor β-1 induces snail transcription factor in epithelial cell lines: mechanisms for epithelial mesenchymal transitions. J. Biol. Chem. 278, 21113–21123 (2003).

    Article  CAS  PubMed  Google Scholar 

  58. Timmerman, L. A. et al. Notch promotes epithelial–mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 18, 99–115 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Nakajima, Y., Yamagishi, T., Hokari, S. & Nakamura, H. Mechanisms involved in valvuloseptal endocardial cushion formation in early cardiogenesis: roles of transforming growth factor (TGF)-β and bone morphogenetic protein (BMP). Anat. Rec. 258, 119–127 (2000).

    Article  CAS  PubMed  Google Scholar 

  60. Zavadil, J., Cermak, L., Soto-Nieves, N. & Bottinger, E. P. Integration of TGF-β/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO J. 23, 1155–1165 (2004). A well-designed study to investigate a complex network of interactions in EMT.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Woodley, D. T. Reepithelialization. in The molecular and cellular biology of wound healing (ed Clarke, A. F.) 339–354 (Plenum Press, New York, 1998).

    Google Scholar 

  62. Martin, P. Wound healing — aiming for perfect skin regeneration. Science 276, 75–81 (1997).

    Article  CAS  PubMed  Google Scholar 

  63. Savagner, P. et al. Developmental transcription factor slug is required for effective re-epithelialization by adult keratinocytes. J. Cell Physiol. 202, 858–866 (2005).

    Article  CAS  PubMed  Google Scholar 

  64. Liu, Y. Epithelial to mesenchymal transition in renal fibrogenesis: pathologic significance, molecular mechanism, and therapeutic intervention. J. Am. Soc. Nephrol. 15, 1–12 (2004).

    Article  CAS  PubMed  Google Scholar 

  65. Kalluri, R. & Neilson, E. G. Epithelial–mesenchymal transition and its implications for fibrosis. J. Clin. Invest. 112, 1776–1784 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Strutz, F. et al. Identification and characterization of a fibroblast marker: FSP1. J. Cell Biol. 130, 393–405 (1995).

    Article  CAS  PubMed  Google Scholar 

  67. Ng, Y. Y. et al. Tubular epithelial-myofibroblast transdifferentiation in progressive tubulointerstitial fibrosis in 5/6 nephrectomized rats. Kidney Int. 54, 864–876 (1998).

    Article  CAS  PubMed  Google Scholar 

  68. Yang, J. & Liu, Y. Dissection of key events in tubular epithelial to myofibroblast transition and its implications in renal interstitial fibrosis. Am. J. Pathol. 159, 1465–1475 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Jinde, K. et al. Tubular phenotypic change in progressive tubulointerstitial fibrosis in human glomerulonephritis. Am. J. Kidney Dis. 38, 761–769 (2001).

    Article  CAS  PubMed  Google Scholar 

  70. Rastaldi, M. P. et al. Epithelial–mesenchymal transition of tubular epithelial cells in human renal biopsies. Kidney Int. 62, 137–146 (2002).

    Article  PubMed  Google Scholar 

  71. Iwano, M. et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J. Clin. Invest. 110, 341–350 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Yang, J. et al. Disruption of tissue-type plasminogen activator gene in mice reduces renal interstitial fibrosis in obstructive nephropathy. J. Clin. Invest. 110, 1525–1538 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Yanez-Mo, M. et al. Peritoneal dialysis and epithelial-to-mesenchymal transition of mesothelial cells. N. Engl. J. Med. 348, 403–413 (2003).

    Article  PubMed  Google Scholar 

  74. Aguilera, A., Yanez-Mo, M., Selgas, R., Sanchez-Madrid, F. & Lopez-Cabrera, M. Epithelial to mesenchymal transition as a triggering factor of peritoneal membrane fibrosis and angiogenesis in peritoneal dialysis patients. Curr. Opin. Investig. Drugs 6, 262–268 (2005).

    CAS  PubMed  Google Scholar 

  75. Margetts, P. J. et al. Transient overexpression of TGF-β1 induces epithelial mesenchymal transition in the rodent peritoneum. J. Am. Soc. Nephrol. 16, 425–436 (2005).

    Article  CAS  PubMed  Google Scholar 

  76. Salez, F. et al. Transforming growth factor-β1 in sarcoidosis. Eur. Respir. J. 12, 913–919 (1998).

    Article  CAS  PubMed  Google Scholar 

  77. Khalil, N. et al. Regulation of the effects of TGF-β1 by activation of latent TGF-β1 and differential expression of TGF-β receptors (TβR-I and TβR-II) in idiopathic pulmonary fibrosis. Thorax 56, 907–915 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Yao, H. W., Xie, Q. M., Chen, J. Q., Deng, Y. M. & Tang, H. F. TGF-β1 induces alveolar epithelial to mesenchymal transition in vitro. Life Sci. 76, 29–37 (2004).

    Article  CAS  PubMed  Google Scholar 

  79. Willis, B. C. et al. Induction of epithelial–mesenchymal transition in alveolar epithelial cells by transforming growth factor-β1: potential role in idiopathic pulmonary fibrosis. Am. J. Pathol. 166, 1321–1332 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. de Iongh, R. U., Wederell, E., Lovicu, F. J. & McAvoy, J. W. Transforming growth factor-β-induced epithelial–mesenchymal transition in the lens: a model for cataract formation. Cells Tissues Organs 179, 43–55 (2005).

    Article  CAS  PubMed  Google Scholar 

  81. Flanders, K. C. Smad3 as a mediator of the fibrotic response. Int. J. Exp. Pathol. 85, 47–64 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Saika, S. et al. Smad3 signaling is required for epithelial–mesenchymal transition of lens epithelium after injury. Am. J. Pathol. 164, 651–663 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Saika, S. et al. Smad3 is required for dedifferentiation of retinal pigment epithelium following retinal detachment in mice. Lab. Invest. 84, 1245–1258 (2004).

    Article  CAS  PubMed  Google Scholar 

  84. Sleeman, J. P. The lymph node as a bridgehead in the metastatic dissemination of tumors. Recent Results Cancer Res. 157, 55–81 (2000).

    Article  CAS  PubMed  Google Scholar 

  85. Tarin, D., Thompson, E. W. & Newgreen, D. F. The fallacy of epithelial mesenchymal transition in neoplasia. Cancer Res. 65, 5996–6000 (2005).

    Article  CAS  PubMed  Google Scholar 

  86. De Craene, B. et al. The transcription factor snail induces tumor cell invasion through modulation of the epithelial cell differentiation program. Cancer Res. 65, 6237–6244 (2005).

    Article  CAS  PubMed  Google Scholar 

  87. Affolter, M. et al. Tube or not tube: remodeling epithelial tissues by branching morphogenesis. Dev. Cell 4, 11–18 (2003).

    Article  CAS  PubMed  Google Scholar 

  88. Nelson, C. M. & Bissell, M. J. Modeling dynamic reciprocity: engineering three-dimensional culture models of breast architecture, function, and neoplastic transformation. Semin. Cancer Biol. 15, 342–352 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  89. O'Brien, P. M. et al. Immunoglobulin genes expressed by B-lymphocytes infiltrating cervical carcinomas show evidence of antigen-driven selection. Cancer Immunol. Immunother. 50, 523–532 (2001).

    Article  CAS  PubMed  Google Scholar 

  90. Debnath, J. & Brugge, J. S. Modelling glandular epithelial cancers in three-dimensional cultures. Nature Rev. Cancer 5, 675–688 (2005).

    Article  CAS  Google Scholar 

  91. Savagner, P., Valles, A. M., Jouanneau, J., Yamada, K. M. & Thiery, J. P. Alternative splicing in fibroblast growth factor receptor 2 is associated with induced epithelial–mesenchymal transition in rat bladder carcinoma cells. Mol. Biol. Cell 5, 851–862 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Pietri, T. et al. Conditional β1-integrin gene deletion in neural crest cells causes severe developmental alterations of the peripheral nervous system. Development 131, 3871–3883 (2004).

    Article  CAS  PubMed  Google Scholar 

  93. Moody, S. E. et al. The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer Cell 8, 197–209 (2005). A remarkable model for the role of EMT in breast cancer progression.

    Article  CAS  PubMed  Google Scholar 

  94. Petersen, O. W. et al. Epithelial to mesenchymal transition in human breast cancer can provide a nonmalignant stroma. Am. J. Pathol. 162, 391–402 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Xue, C., Plieth, D., Venkov, C., Xu, C. & Neilson, E. G. The gatekeeper effect of epithelial–mesenchymal transition regulates the frequency of breast cancer metastasis. Cancer Res. 63, 3386–3394 (2003).

    CAS  PubMed  Google Scholar 

  96. Perl, A. K., Wilgenbus, P., Dahl, U., Semb, H. & Christofori, G. A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature 392, 190–193 (1998).

    Article  CAS  PubMed  Google Scholar 

  97. Dhawan, P. et al. Claudin-1 regulates cellular transformation and metastatic behavior in colon cancer. J. Clin. Invest. 115, 1765–1776 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Huber, M. A. et al. NF-κB is essential for epithelial–mesenchymal transition and metastasis in a model of breast cancer progression. J. Clin. Invest. 114, 569–581 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Yang, J. et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117, 927–939 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported, in part, by the European Union under the auspices of the European Economic Comunity Framework Programme 6 Specific Targeted Research Project BRECOSM (Breast Cancer Metastasis). This review is dedicated to the memory of Professor Shoichiro Tsukita of Kyoto University who passed away in December 2005.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

UniProtKB

FAK

PAR3

Glossary

Mesoderm

In animals with three tissue layers, the mesoderm is the middle layer of tissue, lying between the ectoderm and the endoderm. In vertebrates, it forms the skeleton, muscles, heart, spleen, kidney and other internal organs.

Endoderm

The innermost germ layer of the developing embryo. It gives rise to the lungs, digestive tract, thyroid, thymus, liver and pancreas.

Phyla

Large groups of species that share the same body plan. The animal kingdom is composed of about 30 phyla including Porifera, Cnidaria, Arthropoda, Echinodermata, and Chordata, which includes the Vertebrata as a subphylum.

Porifera

The most primitive phylum of the animal kingdom, it includes sponges.

Cnidaria

Radially symmetrical animals that form a phylum that includes jellyfish, corals, hydra and anemonies.

Blastula stage

An early-stage embryo that is composed of a hollow ball of cells.

Primitive colonial protozoans

Single-celled organisms that live in colonies — they might be the organisms from which Porifera developed.

Diploblastic

Animals that are composed of two cell layers. They belong to the phylum Cnidaria.

Ectoderm

The outermost of the three primary germ layers of the embryo, from which the skin, nerve tissue and sensory organs develop.

Mesenchyme

Embryonic tissue that is composed of loosely organized, unpolarized cells of both mesodermal and ectodermal (for example, neural crest) origin, with a component-rich extracellular matrix.

Organizer

A small dorsal region of the vertebrate gastrula-stage embryo that has the remarkable capacity to organize a complete embryonic body plan. Hilde Mangold and Hans Spemann first identified the organizer in amphibian embryos using tissue transplantation.

Mesendoderm

Cells that form early during gastrulation in the vertebrate and are destined to give rise to mesodermal and endodermal derivatives.

Rostrocaudal

The anterior–posterior (head to tail) polarity of animals.

Neural crest

A transient embryonic structure of vertebrates that appears in the ectoderm at the junction between the neural plate and lateral ectoderm. This structure gives rise to many distinct derivatives following precise migratory routes at each axial level. The derivatives include cranio–facial structures (cartilage, bone, muscles), melanocytes, adrenal medulla, and cells of the sensory and autonomic nervous systems.

Basement membrane

An extracellular-matrix structure that can be visualized by light microscopy and lines the basal side of epithelia.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Thiery, J., Sleeman, J. Complex networks orchestrate epithelial–mesenchymal transitions. Nat Rev Mol Cell Biol 7, 131–142 (2006). https://doi.org/10.1038/nrm1835

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm1835

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing